1
|
Voitovich I, Ty N, Couderc M, Moreau E, Peyrode C, Weber V. Design, synthesis and in vitro evaluation of non-steroidal anti-inflammatory prodrugs for osteoarthritis. Bioorg Chem 2025; 160:108410. [PMID: 40220712 DOI: 10.1016/j.bioorg.2025.108410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/03/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025]
Abstract
Osteoarthritis (OA), a degenerative joint disease characterized by chronic pain and stiffness, is the most common cause of disability in older adults. To date, OA lacks curative treatment and medical care is limited to symptom relief particularly through the use of oral nonsteroidal anti-inflammatory drugs (NSAIDs). However, gastrointestinal and cardiovascular adverse effects and only limited benefits in long-term relief of pain are still associated. Moreover, efficiency is in part impeded by rapid clearance of the drugs and by the special physiological environment of the joint impeding deep penetration of drugs. Hence, to overcome those limitations and improve patient outcome, developing new therapeutic approaches based on anti-inflammatory prodrugs with prolonged drug residence time within the joints appears as a promising strategy in OA. We report herein the development of NSAID prodrugs, derived from diclofenac and naproxen, bearing a positively charged quaternary ammonium (QA) to target the negative-fixed charge density in cartilage by the mean of electrostatic interactions. Our charge-based targeted approach aims to extend the residence time of NSAID within the cartilaginous tissues, leading to a potential decrease of side effects and improved efficiency of locally released drugs. Syntheses of various amide and esters prodrugs of diclofenac and naproxen bearing a QA function were performed, including some hypoxia-activated prodrugs. Since most diclofenac derivatives suffered from high instability preventing any further development, we focused on the naproxen derivatives that were relatively stable in PBS buffer over a 24-h period even if three different degradation patterns were observed in murine plasma. A preliminary screening of their in vitro anti-inflammatory efficacy highlighted a correlation between the PGE-2 inhibition and these cleavage patterns. These results support further in vitro and in vivo evaluations of four of these derivatives in OA models.
Collapse
Affiliation(s)
- Iuliia Voitovich
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Nancy Ty
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Marion Couderc
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France; Rheumatology Department, Gabriel Montpied University Hospital, Clermont-Ferrand, France
| | - Emmanuel Moreau
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Caroline Peyrode
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Valérie Weber
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| |
Collapse
|
2
|
Regeni I, Bonnet S. Supramolecular approaches for the treatment of hypoxic regions in tumours. Nat Rev Chem 2025:10.1038/s41570-025-00705-7. [PMID: 40185999 DOI: 10.1038/s41570-025-00705-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 04/07/2025]
Abstract
Supramolecular chemistry provides a range of 'weak' intermolecular interactions that allow drugs and prodrugs to self-assemble. In the complex biological setting of blood and tumours, these interactions must be stable enough for efficient and selective drug delivery to the tumour site, but weak enough to allow the release of the cytotoxic load. The non-covalent nature of supramolecular interactions enables the detachment of smaller (pro)drug monomers that can penetrate cancer cells differently to the original nanoparticles. Hypoxic tumours show low oxygen levels due to poor vascularization, which poses challenges for drug delivery and generates biological resistances. Supramolecular building blocks specifically designed for hypoxic tumours offer targeted activation of prodrug self-assemblies, enhancing effectiveness against hypoxic cancer cells and hypoxic regions in tumours. This Review explores how supramolecular chemistry can improve (pro)drug delivery and activation in hypoxic tumours.
Collapse
Affiliation(s)
- Irene Regeni
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands.
| | - Sylvestre Bonnet
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
3
|
Sobti A, Skinner H, Wilke CT. Predictors of Radiation Resistance and Novel Radiation Sensitizers in Head and Neck Cancers: Advancing Radiotherapy Efficacy. Semin Radiat Oncol 2025; 35:224-242. [PMID: 40090749 DOI: 10.1016/j.semradonc.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 03/18/2025]
Abstract
Radiation resistance in head and neck squamous cell carcinoma (HNSCC), driven by intrinsic and extrinsic factors, poses a significant challenge in radiation oncology. The key contributors are tumor hypoxia, cancer stem cells, cell cycle checkpoint activation, and DNA repair processes (homologous recombination and non-homologous end-joining). Genetic modifications such as TP53 mutations, KRAS mutations, EGFR overexpression, and abnormalities in DNA repair proteins like BRCA1/2 additionally affect radiation sensitivity. Novel radiosensitizers targeting these pathways demonstrate the potential to overcome resistance. Hypoxia-activated drugs and gold nanoparticles enhance the efficacy of radiotherapy and facilitate targeted distribution. Integrating immunotherapy, especially immune checkpoint inhibitors, with radiation therapy, enhances anti-tumor responses and reduces resistance. Epigenetic alterations, such as DNA methylation and histone acetylation, significantly influence radiation response, with the potential for sensitization through histone deacetylase inhibitors and non-coding RNA regulators. Metabolic changes linked to glucose, lipid, and glutamine metabolism influence radiosensitivity, uncovering new targets for radiosensitization. Human papillomavirus (HPV)-associated malignancies exhibit increased radiosensitivity relative to other tumors due to impaired DNA repair mechanisms and heightened immunogenicity. Furthermore, understanding the interplay between HPV oncoproteins and p53 functionality can enhance treatment strategies for HPV-related cancers. Using DNA damage response inhibitors (PARP, ATM/ATR), cell cycle checkpoint inhibitors (WEE1, CHK1/2), and hypoxia-targeted agents as radiosensitizing strategies exhibit considerable promise. Immunomodulatory approaches, including PD-1 and CTLA-4 inhibitors in conjunction with radiation, enhance anti-tumor immunity. Future directions emphasize personalized radiation therapy using genetics, sophisticated medication delivery systems, adaptive radiotherapy, and real-time monitoring. These integrated strategies seek to diminish radiation resistance and improve therapeutic efficacy in HNSCC.
Collapse
Affiliation(s)
- Aastha Sobti
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | - Heath Skinner
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | - Christopher T Wilke
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA..
| |
Collapse
|
4
|
Wang Q, Suo Y, Tian X. 5-Aminolaevulinic Acid-Mediated Photodynamic Therapy Combined with Tirapazamine Enhances Efficacy in Ovarian Cancer. Biomedicines 2025; 13:724. [PMID: 40149700 PMCID: PMC11939993 DOI: 10.3390/biomedicines13030724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
Objectives: Ovarian cancer is a common gynaecological malignancy. Photodynamic therapy (PDT) mediated by 5-aminolaevulinic acid (5-ALA-PDT) is widely used in clinical practice. However, hypoxia may impact the efficacy of this treatment. In the present study, we combined the bioreductively active drug tirapazamine (TPZ) with PDT to explore its potential in enhancing ovarian cancer cell death. Methods: A cell counting kit-8 assay was used to determine cytotoxicity under different intervention conditions. The distribution of protoporphyrin IX, a metabolite of 5-ALA, was observed using in vivo fluorescence imaging. The effect of the combined treatment was assessed by measuring changes in tumour size following the corresponding interventions and by haematoxylin and eosin staining of tumour tissues. Immunohistochemical staining was used to detect the expression levels of relevant proteins. Results: TPZ exhibited no cytotoxicity under normoxic conditions but was activated under hypoxic conditions, inducing cytotoxic effects that were enhanced when combined with PDT. Over time, protoporphyrin IX achieved systemic distribution, and high drug concentrations were maintained within the tumour. The combination therapy suppressed tumour growth, and pathological staining showed that necrotic tumour areas were significantly enlarged after treatment. The enhanced therapeutic effect may be attributable to the inhibition of the hypoxia-inducible factor-1α/vascular endothelial growth factor axis and PI3K/Akt/mTOR pathway. Conclusions: 5-ALA-PDT combined with TPZ can overcome both the hypoxic state of ovarian cancer tissues and the increased hypoxia induced by PDT, thereby inhibiting tumour growth.
Collapse
Affiliation(s)
- Qian Wang
- Fifth Clinical Medical College, Shanxi Medical University, Taiyuan 030012, China; (Q.W.); (X.T.)
| | - Yuping Suo
- Fifth Clinical Medical College, Shanxi Medical University, Taiyuan 030012, China; (Q.W.); (X.T.)
- Department of Gynaecology and Obstetrics, Shanxi Provincial People’s Hospital, Taiyuan 030012, China
| | - Xiaojuan Tian
- Fifth Clinical Medical College, Shanxi Medical University, Taiyuan 030012, China; (Q.W.); (X.T.)
| |
Collapse
|
5
|
Wang F, Song L, Xu Q, Jia A, Meng X, Jiang H, Zhang R. Hypoxia-selective prodrug restrains tumor cells through triggering mitophagy and inducing apoptosis. Eur J Med Chem 2025; 283:117155. [PMID: 39657461 DOI: 10.1016/j.ejmech.2024.117155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/04/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Hypoxia is a common feature of various solid tumors, which reduces the sensitivity of tumor cells to both radiotherapy and chemotherapy. However, hypoxia also presents an opportunity for tumor-selective therapy. The prodrug strategy, leveraging the hypoxic nature of the tumor microenvironment, shows significant potential for clinical application. Here we present CHD-1, a hypoxia-activated antitumor prodrug that activates in hypoxic environments, effectively inhibiting hypoxic tumor cells while exhibiting no toxicity to normoxic cells. CHD-1 impairs mitochondrial morphology and membrane potential of hypoxic tumor cells, further triggers excessive mitophagy and induces apoptosis. Moreover, prodrug CHD-1 significantly inhibits HeLa xenograft growth in vivo, and shows lower toxicity than parent molecule in an acute toxicity assessment in animal models. This study introduces a promising hypoxia-activated antitumor prodrug with strong potential for further development in hypoxic tumor therapy.
Collapse
Affiliation(s)
- Fangjie Wang
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou, Henan, 450018, China
| | - Lairong Song
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100071, China
| | - Qianqian Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Ang Jia
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Xiangwei Meng
- Department of Drug Clinical Trials, Zibo Central Hospital, Zibo, 255036, China.
| | - Hongfei Jiang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Renshuai Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
6
|
Saha S, Ghosh S, Ghosh S, Nandi S, Nayak A. Unraveling the complexities of colorectal cancer and its promising therapies - An updated review. Int Immunopharmacol 2024; 143:113325. [PMID: 39405944 DOI: 10.1016/j.intimp.2024.113325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Colorectal cancer (CRC) continues to be a global health concern, necessitating further research into its complex biology and innovative treatment approaches. The etiology, pathogenesis, diagnosis, and treatment of colorectal cancer are summarized in this thorough review along with recent developments. The multifactorial nature of colorectal cancer is examined, including genetic predispositions, environmental factors, and lifestyle decisions. The focus is on deciphering the complex interactions between signaling pathways such as Wnt/β-catenin, MAPK, TGF-β as well as PI3K/AKT that participate in the onset, growth, and metastasis of CRC. There is a discussion of various diagnostic modalities that span from traditional colonoscopy to sophisticated molecular techniques like liquid biopsy and radiomics, emphasizing their functions in early identification, prognostication, and treatment stratification. The potential of artificial intelligence as well as machine learning algorithms in improving accuracy as well as efficiency in colorectal cancer diagnosis and management is also explored. Regarding therapy, the review provides a thorough overview of well-known treatments like radiation, chemotherapy, and surgery as well as delves into the newly-emerging areas of targeted therapies as well as immunotherapies. Immune checkpoint inhibitors as well as other molecularly targeted treatments, such as anti-epidermal growth factor receptor (anti-EGFR) as well as anti-vascular endothelial growth factor (anti-VEGF) monoclonal antibodies, show promise in improving the prognosis of colorectal cancer patients, in particular, those suffering from metastatic disease. This review focuses on giving readers a thorough understanding of colorectal cancer by considering its complexities, the present status of treatment, and potential future paths for therapeutic interventions. Through unraveling the intricate web of this disease, we can develop a more tailored and effective approach to treating CRC.
Collapse
Affiliation(s)
- Sayan Saha
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Shreya Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Suman Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Sumit Nandi
- Department of Pharmacology, Gupta College of Technological Sciences, Asansol, West Bengal 713301, India
| | - Aditi Nayak
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India.
| |
Collapse
|
7
|
Inam A, Zhang S, Zhang S, Wu D. AQ4N nanocomposites for hypoxia-associated tumor combination therapy. Biomater Sci 2024; 12:5883-5911. [PMID: 39431892 DOI: 10.1039/d4bm00883a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Hypoxia in solid tumors increases their invasiveness and resistance to therapy, presenting a formidable obstacle in tumor therapy. The hypoxia prodrug banoxantrone (AQ4N) undergoes conversion into its topoisomerase II inhibitor form AQ4 under hypoxic conditions, which inhibits tumor cells while leaving normal cells unharmed. Numerous studies have found that AQ4N significantly enhances the tumor effect while minimizing toxicity to normal tissues when combined with other drugs or therapeutic approaches. Thus, to maximize AQ4N's effectiveness, co-delivery of AQ4N with other therapeutic agents to the tumor site is paramount, leading to the development of multifunctional multicomponent AQ4N nanocomposites thereby emerging as promising candidates for combination therapy in tumor treatment. However, currently there is a lack of systematic analysis and reviews focusing on AQ4N. Herein, this review provides a comprehensive retrospect and analysis of the recent advancements in AQ4N nanocomposites. Specifically, we discuss the synergistic effects observed when AQ4N is combined with chemotherapeutic drugs, radiotherapy, phototherapy, starvation, sonodynamic therapy and immunotherapy in preclinical models. Moreover, the advantages, limitations, and future perspectives of different AQ4N nanocomposites are highlighted, providing researchers from diverse fields with novel insights into tumor treatment.
Collapse
Affiliation(s)
- Amrah Inam
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China.
| | - Shuo Zhang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China.
| | - Shuai Zhang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China.
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China.
| |
Collapse
|
8
|
Lee LC, Lo KK. Leveraging the Photofunctions of Transition Metal Complexes for the Design of Innovative Phototherapeutics. SMALL METHODS 2024; 8:e2400563. [PMID: 39319499 PMCID: PMC11579581 DOI: 10.1002/smtd.202400563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/03/2024] [Indexed: 09/26/2024]
Abstract
Despite the advent of various medical interventions for cancer treatment, the disease continues to pose a formidable global health challenge, necessitating the development of new therapeutic approaches for more effective treatment outcomes. Photodynamic therapy (PDT), which utilizes light to activate a photosensitizer to produce cytotoxic reactive oxygen species (ROS) for eradicating cancer cells, has emerged as a promising approach for cancer treatment due to its high spatiotemporal precision and minimal invasiveness. However, the widespread clinical use of PDT faces several challenges, including the inefficient production of ROS in the hypoxic tumor microenvironment, the limited penetration depth of light in biological tissues, and the inadequate accumulation of photosensitizers at the tumor site. Over the past decade, there has been increasing interest in the utilization of photofunctional transition metal complexes as photosensitizers for PDT applications due to their intriguing photophysical and photochemical properties. This review provides an overview of the current design strategies used in the development of transition metal complexes as innovative phototherapeutics, aiming to address the limitations associated with PDT and achieve more effective treatment outcomes. The current challenges and future perspectives on the clinical translation of transition metal complexes are also discussed.
Collapse
Affiliation(s)
- Lawrence Cho‐Cheung Lee
- Department of ChemistryCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
| | - Kenneth Kam‐Wing Lo
- Department of ChemistryCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
- State Key Laboratory of Terahertz and Millimeter WavesCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
| |
Collapse
|
9
|
Wang Q, Suo Y, Shi R, Wang Y. Studies related to the enhanced the effect of 5-aminolevulinic acid-based photodynamic therapy combined with tirapazamine. Photodiagnosis Photodyn Ther 2024; 49:104287. [PMID: 39059759 DOI: 10.1016/j.pdpdt.2024.104287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
OBJECTIVE 5-aminolevulinic acid (5-ALA) is a precursor of the photosensitizer Protoporphyrin IX (PpIX) and photodynamic therapy (PDT) with 5-ALA has been used in clinical practice. However, tumor cellular hypoxia severely affects the efficiency of photodynamic therapy. In this study, photodynamic therapy was combined with tirapazamine to investigate the effects of the combined intervention and the related mechanisms it may involve. METHODS Colony formation assays were used to demonstrate cell proliferation. Transwell assays were performed to observe the effect on cell invasion and metastasis after the corresponding intervention. DCFH-DA probe was used to detect the reactive oxygen species content. Flow cytometry was used to detect the effects of the interventions on apoptosis and cell cycle. The relevant pathways that may be involved are explored by examining the expression levels of the relevant proteins and genes. RESULTS Colony formation assays indicated that the combined intervention inhibited cell proliferation. Transwell assays demonstrated that PDT combined with TPZ effectively inhibited tumor cell invasion and metastasis. In addition, fluorescence intensity generated by DCFH-DA oxidation was detected indicating that the combined intervention increased the formation of reactive oxygen species. Flow cytometry clearly showed that the combination of PDT and TPZ further increased apoptosis and cell cycle arrest. The results of western blotting and qRT-PCR experiments confirmed that the combination therapy inhibited HIF-1α/VEGF axis and the PI3K/Akt/mTOR pathway activation. CONCLUSION 5-ALA-PDT combined with TPZ can inhibit cell proliferation, increase apoptosis, and inhibit the PI3K/Akt/mTOR pathway, thus inhibiting tumor growth and metastasis and improving anti-cancer effects.
Collapse
Affiliation(s)
- Qian Wang
- Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, 030012, Shanxi, China
| | - Yuping Suo
- Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, 030012, Shanxi, China; Department of Gynaecology and Obstetrics, Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, China.
| | - Rui Shi
- Department of Gynaecology and Obstetrics, Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, China
| | - Yulan Wang
- Department of Gynaecology and Obstetrics, Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, China
| |
Collapse
|
10
|
Lee LCC, Lo KKW. Shining New Light on Biological Systems: Luminescent Transition Metal Complexes for Bioimaging and Biosensing Applications. Chem Rev 2024; 124:8825-9014. [PMID: 39052606 PMCID: PMC11328004 DOI: 10.1021/acs.chemrev.3c00629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Luminescence imaging is a powerful and versatile technique for investigating cell physiology and pathology in living systems, making significant contributions to life science research and clinical diagnosis. In recent years, luminescent transition metal complexes have gained significant attention for diagnostic and therapeutic applications due to their unique photophysical and photochemical properties. In this Review, we provide a comprehensive overview of the recent development of luminescent transition metal complexes for bioimaging and biosensing applications, with a focus on transition metal centers with a d6, d8, and d10 electronic configuration. We elucidate the structure-property relationships of luminescent transition metal complexes, exploring how their structural characteristics can be manipulated to control their biological behavior such as cellular uptake, localization, biocompatibility, pharmacokinetics, and biodistribution. Furthermore, we introduce the various design strategies that leverage the interesting photophysical properties of luminescent transition metal complexes for a wide variety of biological applications, including autofluorescence-free imaging, multimodal imaging, organelle imaging, biological sensing, microenvironment monitoring, bioorthogonal labeling, bacterial imaging, and cell viability assessment. Finally, we provide insights into the challenges and perspectives of luminescent transition metal complexes for bioimaging and biosensing applications, as well as their use in disease diagnosis and treatment evaluation.
Collapse
Affiliation(s)
- Lawrence Cho-Cheung Lee
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Units 1503-1511, 15/F, Building 17W, Hong Kong Science Park, New Territories, Hong Kong, P. R. China
| | - Kenneth Kam-Wing Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
- State Key Laboratory of Terahertz and Millimeter Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| |
Collapse
|
11
|
Anichina K, Lumov N, Bakov V, Yancheva D, Georgiev N. Recent Advances in the Application of Nitro(het)aromatic Compounds for Treating and/or Fluorescent Imaging of Tumor Hypoxia. Molecules 2024; 29:3475. [PMID: 39124883 PMCID: PMC11314162 DOI: 10.3390/molecules29153475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
This review delves into recent advancements in the field of nitro(het)aromatic bioreductive agents tailored for hypoxic environments. These compounds are designed to exploit the low-oxygen conditions typically found in solid tumors, making them promising candidates for targeted cancer therapies. Initially, this review focused on their role as gene-directed enzyme prodrugs, which are inert until activated by specific enzymes within tumor cells. Upon activation, these prodrugs undergo chemical transformations that convert them into potent cytotoxic agents, selectively targeting cancerous tissue while sparing healthy cells. Additionally, this review discusses recent developments in prodrug conjugates containing nitro(het)aromatic moieties, designed to activate under low-oxygen conditions within tumors. This approach enhances their efficacy and specificity in cancer treatment. Furthermore, this review covers innovative research on using nitro(het)aromatic compounds as fluorescent probes for imaging hypoxic tumors. These probes enable non-invasive visualization of low-oxygen regions within tumors, providing valuable insights for the diagnosis, treatment planning, and monitoring of therapeutic responses. We hope this review will inspire researchers to design and synthesize improved compounds for selective cancer treatment and early diagnostics.
Collapse
Affiliation(s)
- Kameliya Anichina
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria; (K.A.); (N.L.); (V.B.); (D.Y.)
| | - Nikolay Lumov
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria; (K.A.); (N.L.); (V.B.); (D.Y.)
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev str. Bl. 9, 1113 Sofia, Bulgaria
| | - Ventsislav Bakov
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria; (K.A.); (N.L.); (V.B.); (D.Y.)
| | - Denitsa Yancheva
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria; (K.A.); (N.L.); (V.B.); (D.Y.)
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev str. Bl. 9, 1113 Sofia, Bulgaria
| | - Nikolai Georgiev
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria; (K.A.); (N.L.); (V.B.); (D.Y.)
| |
Collapse
|
12
|
Jagathesan K, Roy S. Recent Development of Transition Metal Complexes as Chemotherapeutic Hypoxia Activated Prodrug (HAP). ChemMedChem 2024; 19:e202400127. [PMID: 38634306 DOI: 10.1002/cmdc.202400127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 04/19/2024]
Abstract
Hypoxia is a state characterized by low concentration of Oxygen. Hypoxic state is often found in the central region of solid tumors. Hypoxia is associated with abnormal neovascularization resulted in poor blood flow in tissues and increased proliferation of tumor cells, imbalance between O2 supply and O2 consumption in tumor cells, high concentration of proton and strong reducibility. And, these abnormalities enhance the survival potency of the hypoxic tumours and increase the resistance towards chemotherapy and radiotherapy. One of the approach for treating hypoxic region of tumour is to use reducing environment of hypoxic tumours for reducing a molecule (hypoxia activated prodrug, HAP) and as a result the active drug will be released in hypoxic region in a controlled manner from the prodrug and kill the hypoxic tumour. Co(III) and Pt(IV) complexes with monodentate active drug molecule in the axial position can be reduced to Co(II) and Pt(II) moieties and as a result, the axial ligands (active drug) could come out from the metal center and could show its anticancer activity. In this review we have highlighted the research articles where transition metal-based complexes are used as chemotherapeutic hypoxia activated prodrug molecules which are reported in last 5 years.
Collapse
Affiliation(s)
- K Jagathesan
- Dept. of Chemistry, School of Advance Sciences, Vellore Institute of Technology, Vellore, 632014, India
| | - Sovan Roy
- Dept. of Chemistry, School of Advance Sciences, Vellore Institute of Technology, Vellore, 632014, India
| |
Collapse
|
13
|
Tosun Ç, Wallabregue ALD, Mallerman M, Phillips SE, Edwards CM, Conway SJ, Hammond EM. Antibody-Based Imaging of Bioreductive Prodrug Release in Hypoxia. JACS AU 2023; 3:3237-3246. [PMID: 38034969 PMCID: PMC10685431 DOI: 10.1021/jacsau.3c00562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 12/02/2023]
Abstract
Regions of hypoxia occur in most tumors and are a predictor of poor patient prognosis. Hypoxia-activated prodrugs (HAPs) provide an ideal strategy to target the aggressive, hypoxic, fraction of a tumor, while protecting the normal tissue from toxicity. A key challenge associated with the development of novel HAPs, however, is the ability to visualize the delivery of the prodrug to hypoxic regions and determine where it has been activated. Here, we report a modified version of the commonly used nitroimidazole bioreductive group that incorporates the fluoroethyl epitope of the antibody-based hypoxia imaging agent, EF5. Attachment of this group to the red fluorescent dye, dicyanomethylene (DCM), enabled us to correlate the release of the DCM dye with imaging of the reduced bioreductive group using the EF5 antibody. This study confirmed that the antibody was imaging reduction and fragmentation of the pro-fluorophore. We next employed the modified bioreductive group to synthesize a new prodrug of the KDAC inhibitor Panobinostat, EF5-Pano. Release of EF5-Pano in hypoxic multiple myeloma cells was imaged using the EF5 antibody, and the presence of an imaging signal correlated with apoptosis and a reduction in cell viability. Therefore, EF5-Pano is an imageable HAP with a proven cytotoxic effect in multiple myeloma, which could be utilized in future in vivo experiments.
Collapse
Affiliation(s)
- Çağla Tosun
- Department
of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, U.K.
| | - Antoine L. D. Wallabregue
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K.
| | - Maxim Mallerman
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K.
| | - Sarah E. Phillips
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K.
| | - Claire M. Edwards
- Nuffield
Department of Surgical Sciences, University
of Oxford, Oxford OX3 7HE, U.K.
- Nuffield
Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, U.K.
| | - Stuart J. Conway
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K.
- Department
of Chemistry & Biochemistry, University
of California, 607 Charles
E. Young Drive East, Los Angeles, California CA90095, United States
| | - Ester M. Hammond
- Department
of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, U.K.
| |
Collapse
|
14
|
Bhise K, Gavande NS, Iyer AK. Leveraging hypoxia in triple-negative breast cancer as a promising treatment strategy. Drug Discov Today 2023; 28:103761. [PMID: 37660983 DOI: 10.1016/j.drudis.2023.103761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/08/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Current treatment strategies for triple-negative breast cancer (TNBC) are based upon conventional chemotherapy, immunotherapy, or a combination of both. The treatment regimen for chemotherapy is often a combination of two or more drugs, either dose dense or low dose for synergy. Anthracyclines, alkylating agents, antimicrotubule agents, and antimetabolites for early-stage TNBC; and antimetabolites, non-taxane microtubule inhibitors, and cross-linker platinums for late-stage TNBC are usually administered in the clinical setting. Newer options for patients with advanced TNBC, such as poly (ADP-ribose) polymerase (PARP) inhibitors and immune checkpoint inhibitors, have recently emerged for cases where surgery is not a viable option and the disease has metastasized. This review outlines the current trends in hypoxia-inspired treatment strategies for TNBC with a focus on clinical trials.
Collapse
Affiliation(s)
- Ketki Bhise
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
| | - Navnath S Gavande
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA; Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, USA
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA; Molecular Imaging Program, Karmanos Cancer Institute, Detroit, MI, USA.
| |
Collapse
|
15
|
Guo JS, Li JJ, Wang ZH, Liu Y, Yue YX, Li HB, Zhao XH, Sun YJ, Ding YH, Ding F, Guo DS, Wang L, Chen Y. Dual hypoxia-responsive supramolecular complex for cancer target therapy. Nat Commun 2023; 14:5634. [PMID: 37704601 PMCID: PMC10500001 DOI: 10.1038/s41467-023-41388-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 09/01/2023] [Indexed: 09/15/2023] Open
Abstract
The prognosis with pancreatic cancer is among the poorest of any human cancer. One of the important factors is the tumor hypoxia. Targeting tumor hypoxia is considered a desirable therapeutic option. However, it has not been translated into clinical success in the treatment of pancreatic cancer. With enhanced cytotoxicities against hypoxic pancreatic cancer cells, BE-43547A2 (BE) may serve as a promising template for hypoxia target strategy. Here, based on rational modification, a BE prodrug (NMP-BE) is encapsulated into sulfonated azocalix[5]arene (SAC5A) to generate a supramolecular dual hypoxia-responsive complex NMP-BE@SAC5A. Benefited from the selective load release within cancer cells, NMP-BE@SAC5A markedly suppresses tumor growth at low dose in pancreatic cancer cells xenograft murine model without developing systemic toxicity. This research presents a strategy for the modification of covalent compounds to achieve efficient delivery within tumors, a horizon for the realization of safe and reinforced hypoxia target therapy using a simple approach.
Collapse
Affiliation(s)
- Jian-Shuang Guo
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China
| | - Juan-Juan Li
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
| | - Ze-Han Wang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
| | - Yang Liu
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China
| | - Yu-Xin Yue
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
| | - Hua-Bin Li
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
| | - Xiu-He Zhao
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China
| | - Yuan-Jun Sun
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China
| | - Ya-Hui Ding
- College of Chemistry, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Fei Ding
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China
| | - Dong-Sheng Guo
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, 300071, China.
| | - Liang Wang
- College of Chemistry, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.
| | - Yue Chen
- College of Chemistry, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| |
Collapse
|
16
|
Lam KH, Ma S. Noncellular components in the liver cancer stem cell niche: Biology and potential clinical implications. Hepatology 2023; 78:991-1005. [PMID: 35727189 DOI: 10.1002/hep.32629] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 02/06/2023]
Abstract
Cancer stem cells (CSCs) are now recognized as one of the major root causes of therapy failure and tumor recurrence in hepatocellular carcinoma (HCC). Early studies in the field focused primarily on the intrinsic regulators of CSC maintenance, but in recent years, mounting evidence has demonstrated the presence and role of extrinsic regulators in the tumor microenvironment (TME) in the control of liver CSCs. In addition to direct interaction with cellular components, noncellular components, including the extracellular matrix, hypoxia, nutrient deprivation, and secreted molecules within the tumor stroma and hepatitis viruses, also play a critical role in shaping the CSC niche. In this review, we highlight how various noncellular components in the TME play a role in regulating CSCs and how CSCs secrete components to interact with the TME to generate their own niche, working hand in hand to drive tumor physiology in HCC. In addition, we describe the potential clinical applications of these findings and propose perspectives on future research of noncellular components in the liver CSC niche.
Collapse
Affiliation(s)
- Ka-Hei Lam
- School of Biomedical Sciences , Li Ka Shing Faculty of Medicine , The University of Hong Kong , Hong Kong , Hong Kong
| | - Stephanie Ma
- School of Biomedical Sciences , Li Ka Shing Faculty of Medicine , The University of Hong Kong , Hong Kong , Hong Kong
- The University of Hong Kong , Shenzhen Hospital , Hong Kong , Hong Kong
- State Key Laboratory of Liver Research , The University of Hong Kong , Hong Kong , Hong Kong
| |
Collapse
|
17
|
Drzał A, Dziurman G, Hoła P, Lechowski J, Delalande A, Swakoń J, Pichon C, Elas M. Murine Breast Cancer Radiosensitization Using Oxygen Microbubbles and Metformin: Vessels Are the Key. Int J Mol Sci 2023; 24:12156. [PMID: 37569531 PMCID: PMC10418665 DOI: 10.3390/ijms241512156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Radiotherapy is a cornerstone of cancer treatment, but tumor hypoxia and resistance to radiation remain significant challenges. Vascular normalization has emerged as a strategy to improve oxygenation and enhance therapeutic outcomes. In this study, we examine the radiosensitization potential of vascular normalization using metformin, a widely used anti-diabetic drug, and oxygen microbubbles (OMBs). We investigated the synergistic action of metformin and OMBs and the impact of this therapeutic combination on the vasculature, oxygenation, invasiveness, and radiosensitivity of murine 4T1 breast cancer. We employed in vivo Doppler ultrasonographic imaging for vasculature analysis, electron paramagnetic resonance oximetry, and immunohistochemical assessment of microvessels, perfusion, and invasiveness markers. Our findings demonstrate that both two-week metformin therapy and oxygen microbubble treatment normalize abnormal cancer vasculature. The combination of metformin and OMB yielded more pronounced and sustained effects than either treatment alone. The investigated therapy protocols led to nearly twice the radiosensitivity of 4T1 tumors; however, no significant differences in radiosensitivity were observed between the various treatment groups. Despite these improvements, resistance to treatment inevitably emerged, leading to the recurrence of hypoxia and an increased incidence of metastasis.
Collapse
Affiliation(s)
- Agnieszka Drzał
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Jagiellonian University, 30-387 Krakow, Poland; (A.D.); (G.D.); (P.H.); (J.L.)
| | - Gabriela Dziurman
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Jagiellonian University, 30-387 Krakow, Poland; (A.D.); (G.D.); (P.H.); (J.L.)
- Doctoral School of Exact and Natural Sciences, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Jagiellonian University, 30-387 Krakow, Poland
| | - Paweł Hoła
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Jagiellonian University, 30-387 Krakow, Poland; (A.D.); (G.D.); (P.H.); (J.L.)
| | - Jakub Lechowski
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Jagiellonian University, 30-387 Krakow, Poland; (A.D.); (G.D.); (P.H.); (J.L.)
| | - Anthony Delalande
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France; (A.D.); (C.P.)
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
| | - Jan Swakoń
- Institute of Nuclear Physics, Polish Academy of Sciences, 31-342 Krakow, Poland;
| | - Chantal Pichon
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France; (A.D.); (C.P.)
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
- Institut Universitaire de France, 75231 Paris, France
| | - Martyna Elas
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics and Cancer Biology, Jagiellonian University, 30-387 Krakow, Poland; (A.D.); (G.D.); (P.H.); (J.L.)
| |
Collapse
|
18
|
Abi-Jaoudeh N, Sadeghi B, Javan H, Na J, Beaton G, Tucci F, Ravula S, Imagawa DK. Drug-Eluting Embolic Loaded with Tyrosine Kinase Inhibitor Targeted Therapies for Transarterial Chemoembolization in a VX2 Model. Cancers (Basel) 2023; 15:3236. [PMID: 37370846 DOI: 10.3390/cancers15123236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/05/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Drug-eluting embolic transarterial chemoembolization (DEE-TACE) improves the overall survival of hepatocellular carcinoma (HCC), but the agents used are not tailored to HCC. Our patented liposomal formulation enables the loading and elution of targeted therapies onto DEEs. This study aimed to establish the safety, feasibility, and pharmacokinetics of sorafenib or regorafenib DEE-TACE in a VX2 model. DEE-TACE was performed in VX2 hepatic tumors in a selective manner until stasis using liposomal sorafenib- or regorafenib-loaded DEEs. The animals were euthanized at 1, 24, and 72 h timepoints post embolization. Blood samples were taken for pharmacokinetics at 5 and 20 min and at 1, 24, and 72 h. Measurements of sorafenib or regorafenib were performed in all tissue samples on explanted hepatic tissue using the same mass spectrometry method. Histopathological examinations were carried out on tumor tissues and non-embolized hepatic specimens. DEE-TACE was performed on 23 rabbits. The plasma concentrations of sorafenib and regorafenib were statistically significantly several folds lower than the embolized liver at all examined timepoints. This study demonstrates the feasibility of loading sorafenib or regorafenib onto commercially available DEEs for use in TACE. The drugs eluted locally without release into systemic circulation.
Collapse
Affiliation(s)
- Nadine Abi-Jaoudeh
- Department of Radiological Sciences, University of California Irvine, Orange, CA 92697, USA
| | - Ben Sadeghi
- Department of Radiological Sciences, University of California Irvine, Orange, CA 92697, USA
| | - Hanna Javan
- Department of Radiological Sciences, University of California Irvine, Orange, CA 92697, USA
| | - Jim Na
- Cullgen, Inc., San Diego, CA 92130, USA
| | | | - Fabio Tucci
- Epigen Biosciences, San Diego, CA 92121, USA
| | | | - David K Imagawa
- Department of Surgery, University of California Irvine, Orange, CA 92697, USA
| |
Collapse
|
19
|
Huynh KN, Rao S, Roth B, Bryan T, Fernando DM, Dayyani F, Imagawa D, Abi-Jaoudeh N. Targeting Hypoxia-Inducible Factor-1α for the Management of Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:2738. [PMID: 37345074 DOI: 10.3390/cancers15102738] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Hypoxia-inducible factor 1 alpha (HIF-1α) is a transcription factor that regulates the cellular response to hypoxia and is upregulated in all types of solid tumor, leading to tumor angiogenesis, growth, and resistance to therapy. Hepatocellular carcinoma (HCC) is a highly vascular tumor, as well as a hypoxic tumor, due to the liver being a relatively hypoxic environment compared to other organs. Trans-arterial chemoembolization (TACE) and trans-arterial embolization (TAE) are locoregional therapies that are part of the treatment guidelines for HCC but can also exacerbate hypoxia in tumors, as seen with HIF-1α upregulation post-hepatic embolization. Hypoxia-activated prodrugs (HAPs) are a novel class of anticancer agent that are selectively activated under hypoxic conditions, potentially allowing for the targeted treatment of hypoxic HCC. Early studies targeting hypoxia show promising results; however, further research is needed to understand the effects of HAPs in combination with embolization in the treatment of HCC. This review aims to summarize current knowledge on the role of hypoxia and HIF-1α in HCC, as well as the potential of HAPs and liver-directed embolization.
Collapse
Affiliation(s)
- Kenneth N Huynh
- Division of Interventional Radiology, Department of Radiological Sciences, University of California Irvine, Orange, CA 92868, USA
| | - Sriram Rao
- Division of Interventional Radiology, Department of Radiological Sciences, University of California Irvine, Orange, CA 92868, USA
| | - Bradley Roth
- Division of Interventional Radiology, Department of Radiological Sciences, University of California Irvine, Orange, CA 92868, USA
| | - Theodore Bryan
- Division of Interventional Radiology, Department of Radiological Sciences, University of California Irvine, Orange, CA 92868, USA
| | - Dayantha M Fernando
- Division of Interventional Radiology, Department of Radiological Sciences, University of California Irvine, Orange, CA 92868, USA
| | - Farshid Dayyani
- Division of Hematology and Oncology, Department of Medicine, Chao Family Comprehensive Cancer Center, University of California Irvine, Orange, CA 92868, USA
| | - David Imagawa
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, University of California Irvine, Orange, CA 92868, USA
| | - Nadine Abi-Jaoudeh
- Division of Interventional Radiology, Department of Radiological Sciences, University of California Irvine, Orange, CA 92868, USA
| |
Collapse
|
20
|
Smith PJ, McKeown SR, Patterson LH. Targeting DNA topoisomerase IIα (TOP2A) in the hypoxic tumour microenvironment using unidirectional hypoxia-activated prodrugs (uHAPs). IUBMB Life 2023; 75:40-54. [PMID: 35499745 PMCID: PMC10084299 DOI: 10.1002/iub.2619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/24/2022] [Accepted: 04/03/2022] [Indexed: 12/29/2022]
Abstract
The hypoxic tumour microenvironment (hTME), arising from inadequate and chaotic vascularity, can present a major obstacle for the treatment of solid tumours. Hypoxic tumour cells compromise responses to treatment since they can generate resistance to radiotherapy, chemotherapy and immunotherapy. The hTME impairs the delivery of a range of anti-cancer drugs, creates routes for metastasis and exerts selection pressures for aggressive phenotypes; these changes potentially occur within an immunosuppressed environment. Therapeutic strategies aimed at the hTME include targeting the molecular changes associated with hypoxia. An alternative approach is to exploit the prevailing lack of oxygen as a principle for the selective activation of prodrugs to target cellular components within the hTME. This review focuses on the design concepts and rationale for the use of unidirectional Hypoxia-Activated Prodrugs (uHAPs) to target the hTME as exemplified by the uHAPs AQ4N and OCT1002. These agents undergo irreversible reduction in a hypoxic environment to active forms that target DNA topoisomerase IIα (TOP2A). This nuclear enzyme is essential for cell division and is a recognised chemotherapeutic target. An activated uHAP interacts with the enzyme-DNA complex to induce DNA damage, cell cycle arrest and tumour cell death. uHAPs are designed to overcome the shortcomings of conventional HAPs and offer unique pharmacodynamic properties for effective targeting of TOP2A in the hTME. uHAP therapy in combination with standard of care treatments has the potential to enhance outcomes by co-addressing the therapeutic challenge presented by the hTME.
Collapse
Affiliation(s)
- Paul J Smith
- Cancer and Genetics Division, School of Medicine, Cardiff University, Cardiff, UK
| | | | - Laurence H Patterson
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
21
|
Zhao G, Jin Y, Gao S, Xiao T, Fan M, Liu D, Zhang J, Li Z, Zhou X, Liu H. Improving the Therapeutic Efficiency of Hypoxic-Activated Prodrugs by Enhancing Hypoxia in Solid Tumors. ACS Biomater Sci Eng 2022; 8:1604-1612. [PMID: 35348331 DOI: 10.1021/acsbiomaterials.2c00104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The low sensitivity of hypoxic regions in solid tumors to radiotherapy and chemotherapy remains a major obstacle to cancer treatment. By taking advantage of hypoxic-activated prodrugs, tirapazamine (TPZ), generating cytotoxic reductive products and the glucose oxidase (GOx)-based glucose oxidation reaction, we designed a nanodrug-loading system that combined TPZ-induced chemotherapy with GOx-mediated cancer-orchestrated starvation therapy and cancer oxidation therapy. In this work, we first prepared mesoporous silica (MSN) loaded with TPZ. Then, in order to prevent the leakage of TPZ in advance, the surface was coated with a layer of carMOF formed by Fe3+ and carbenicillin (car), and GOx was adsorbed on the outermost layer to form the final nanosystem MSN-TPZ@carMOF-GOx (MT@c-G). GOx could effectively consume oxygen and catalyzed glucose into gluconic acid and hydrogen peroxide. First, the generated gluconic acid lowered the pH of tumor tissues, promoted the decomposition of carMOF, and released TPZ. Second, oxygen consumption could improve the degree of hypoxia in tumor tissues, so that enhanced the activity of TPZ. Furthermore, GOx could generate cancer-orchestrated starvation/oxidation therapy. Therefore, our study provided a new strategy that TPZ combined with GOx achieved starvation/oxidation/chemotherapy for enhancing anticancer effects in hypoxic regions.
Collapse
Affiliation(s)
- Gaoqian Zhao
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China.,Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Yan Jin
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China.,College of Chemistry & Environmental Science, Hebei University, Baoding 071002, China
| | - Shutao Gao
- College of Science, Hebei Agricultural University, Baoding 071002, China
| | - Tingshan Xiao
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China.,Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Miao Fan
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China.,College of Chemistry & Environmental Science, Hebei University, Baoding 071002, China
| | - Dandan Liu
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China.,College of Chemistry & Environmental Science, Hebei University, Baoding 071002, China
| | - Jinchao Zhang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China.,College of Chemistry & Environmental Science, Hebei University, Baoding 071002, China
| | - Zhenhua Li
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan 523059, China
| | - Xiaohan Zhou
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan 523059, China
| | - Huifang Liu
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China.,Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| |
Collapse
|
22
|
Park K, Veena MS, Shin DS. Key Players of the Immunosuppressive Tumor Microenvironment and Emerging Therapeutic Strategies. Front Cell Dev Biol 2022; 10:830208. [PMID: 35345849 PMCID: PMC8957227 DOI: 10.3389/fcell.2022.830208] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/14/2022] [Indexed: 12/14/2022] Open
Abstract
The tumor microenvironment (TME) is a complex, dynamic battlefield for both immune cells and tumor cells. The advent of the immune checkpoint inhibitors (ICI) since 2011, such as the anti-cytotoxic T-lymphocyte associated protein (CTLA)-4 and anti-programmed cell death receptor (PD)-(L)1 antibodies, provided powerful weapons in the arsenal of cancer treatments, demonstrating unprecedented durable responses for patients with many types of advanced cancers. However, the response rate is generally low across tumor types and a substantial number of patients develop acquired resistance. These primary or acquired resistance are attributed to various immunosuppressive elements (soluble and cellular factors) and alternative immune checkpoints in the TME. Therefore, a better understanding of the TME is absolutely essential to develop therapeutic strategies to overcome resistance. Numerous clinical studies are underway using ICIs and additional agents that are tailored to the characteristics of the tumor or the TME. Some of the combination treatments are already approved by the Food and Drug Administration (FDA), such as platinum-doublet chemotherapy, tyrosine kinase inhibitor (TKI) -targeting vascular endothelial growth factor (VEGF) combined with anti-PD-(L)1 antibodies or immuno-immuno combinations (anti-CTLA-4 and anti-PD-1). In this review, we will discuss the key immunosuppressive cells, metabolites, cytokines or chemokines, and hypoxic conditions in the TME that contribute to tumor immune escape and the prospect of relevant clinical trials by targeting these elements in combination with ICIs.
Collapse
Affiliation(s)
- Kevin Park
- Department of Medicine, Division of Hematology/Oncology, Los Angeles, CA, United States.,VA Greater Los Angeles Healthcare System, University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Mysore S Veena
- Department of Medicine, Division of Hematology/Oncology, Los Angeles, CA, United States.,VA Greater Los Angeles Healthcare System, University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Daniel Sanghoon Shin
- Department of Medicine, Division of Hematology/Oncology, Los Angeles, CA, United States.,VA Greater Los Angeles Healthcare System, University of California, Los Angeles (UCLA), Los Angeles, CA, United States.,Molecular Biology Institute, Los Angeles, CA, United States.,Jonsson Comprehensive Cancer Center, Los Angeles, CA, United States
| |
Collapse
|
23
|
Li H, Kim H, Xu F, Han J, Yao Q, Wang J, Pu K, Peng X, Yoon J. Activity-based NIR fluorescent probes based on the versatile hemicyanine scaffold: design strategy, biomedical applications, and outlook. Chem Soc Rev 2022; 51:1795-1835. [PMID: 35142301 DOI: 10.1039/d1cs00307k] [Citation(s) in RCA: 198] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The discovery of a near-infrared (NIR, 650-900 nm) fluorescent chromophore hemicyanine dye with high structural tailorability is of great significance in the field of detection, bioimaging, and medical therapeutic applications. It exhibits many outstanding advantages including absorption and emission in the NIR region, tunable spectral properties, high photostability as well as a large Stokes shift. These properties are superior to those of conventional fluorogens, such as coumarin, fluorescein, naphthalimides, rhodamine, and cyanine. Researchers have made remarkable progress in developing activity-based multifunctional fluorescent probes based on hemicyanine skeletons for monitoring vital biomolecules in living systems through the output of fluorescence/photoacoustic signals, and integration of diagnosis and treatment of diseases using chemotherapy or photothermal/photodynamic therapy or combination therapy. These achievements prompted researchers to develop more smart fluorescent probes using a hemicyanine fluorogen as a template. In this review, we begin by describing the brief history of the discovery of hemicyanine dyes, synthetic approaches, and design strategies for activity-based functional fluorescent probes. Then, many selected hemicyanine-based probes that can detect ions, small biomolecules, overexpressed enzymes and diagnostic reagents for diseases are systematically highlighted. Finally, potential drawbacks and the outlook for future investigation and clinical medicine transformation of hemicyanine-based activatable functional probes are also discussed.
Collapse
Affiliation(s)
- Haidong Li
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China. .,School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Heejeong Kim
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Korea.
| | - Feng Xu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China. .,The Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Jingjing Han
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Korea.
| | - Qichao Yao
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China.
| | - Jingyun Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China. .,School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, 637457, Singapore. .,Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China. .,Research Institute of Dalian University of Technology in Shenzhen, Nanshan District, Shenzhen 518057, China
| | - Juyoung Yoon
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Korea.
| |
Collapse
|
24
|
Lee LCC, Lo KKW. Strategic design of photofunctional transition metal complexes for cancer diagnosis and therapy. ADVANCES IN INORGANIC CHEMISTRY 2022. [DOI: 10.1016/bs.adioch.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
25
|
Habibzadeh Mashatooki M, Ghalami-Choobar B. Improved drug delivery and competitive adsorption of paclitaxel and mitomycin C anticancer drugs on the Boron-nitride nanoparticles: A molecular dynamics insight. Phys Chem Chem Phys 2022; 24:6639-6654. [DOI: 10.1039/d1cp04006e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The competitive aggregated adsorption and molecular interactions between paclitaxel (PX) and mitomycin C (MMC) molecules on the surface of boron nitride nanosheet (BNNS) was investigated using molecular dynamics method. BNNS...
Collapse
|
26
|
van der Wiel AM, Jackson-Patel V, Niemans R, Yaromina A, Liu E, Marcus D, Mowday AM, Lieuwes NG, Biemans R, Lin X, Fu Z, Kumara S, Jochems A, Ashoorzadeh A, Anderson RF, Hicks KO, Bull MR, Abbattista MR, Guise CP, Deschoemaeker S, Thiolloy S, Heyerick A, Solivio MJ, Balbo S, Smaill JB, Theys J, Dubois LJ, Patterson AV, Lambin P. Selectively Targeting Tumor Hypoxia With the Hypoxia-Activated Prodrug CP-506. Mol Cancer Ther 2021; 20:2372-2383. [PMID: 34625504 PMCID: PMC9398139 DOI: 10.1158/1535-7163.mct-21-0406] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/23/2021] [Accepted: 09/30/2021] [Indexed: 01/07/2023]
Abstract
Hypoxia-activated prodrugs (HAP) are a promising class of antineoplastic agents that can selectively eliminate hypoxic tumor cells. This study evaluates the hypoxia-selectivity and antitumor activity of CP-506, a DNA alkylating HAP with favorable pharmacologic properties. Stoichiometry of reduction, one-electron affinity, and back-oxidation rate of CP-506 were characterized by fast-reaction radiolytic methods with observed parameters fulfilling requirements for oxygen-sensitive bioactivation. Net reduction, metabolism, and cytotoxicity of CP-506 were maximally inhibited at oxygen concentrations above 1 μmol/L (0.1% O2). CP-506 demonstrated cytotoxicity selectively in hypoxic 2D and 3D cell cultures with normoxic/anoxic IC50 ratios up to 203. Complete resistance to aerobic (two-electron) metabolism by aldo-keto reductase 1C3 was confirmed through gain-of-function studies while retention of hypoxic (one-electron) bioactivation by various diflavin oxidoreductases was also demonstrated. In vivo, the antitumor effects of CP-506 were selective for hypoxic tumor cells and causally related to tumor oxygenation. CP-506 effectively decreased the hypoxic fraction and inhibited growth of a wide range of hypoxic xenografts. A multivariate regression analysis revealed baseline tumor hypoxia and in vitro sensitivity to CP-506 were significantly correlated with treatment response. Our results demonstrate that CP-506 selectively targets hypoxic tumor cells and has broad antitumor activity. Our data indicate that tumor hypoxia and cellular sensitivity to CP-506 are strong determinants of the antitumor effects of CP-506.
Collapse
Affiliation(s)
- Alexander M.A. van der Wiel
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW – School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Victoria Jackson-Patel
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Raymon Niemans
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW – School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Ala Yaromina
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW – School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Emily Liu
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Damiënne Marcus
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW – School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Alexandra M. Mowday
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW – School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands.,Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Natasja G. Lieuwes
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW – School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Rianne Biemans
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW – School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Xiaojing Lin
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Zhe Fu
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Sisira Kumara
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Arthur Jochems
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW – School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Amir Ashoorzadeh
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Robert F. Anderson
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Kevin O. Hicks
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Matthew R. Bull
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Maria R. Abbattista
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Christopher P. Guise
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | | | | | | | | | - Silvia Balbo
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Jeff B. Smaill
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Jan Theys
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW – School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Ludwig J. Dubois
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW – School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Adam V. Patterson
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,Corresponding Author: Adam V. Patterson, Auckland Cancer Society Research Centre, University of Auckland, Faculty of Medicine and Health Sciences, Auckland 1142, New Zealand. E-mail:
| | - Philippe Lambin
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW – School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
27
|
Vilaplana-Lopera N, Besh M, Moon EJ. Targeting Hypoxia: Revival of Old Remedies. Biomolecules 2021; 11:1604. [PMID: 34827602 PMCID: PMC8615589 DOI: 10.3390/biom11111604] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/14/2022] Open
Abstract
Tumour hypoxia is significantly correlated with patient survival and treatment outcomes. At the molecular level, hypoxia is a major driving factor for tumour progression and aggressiveness. Despite the accumulative scientific and clinical efforts to target hypoxia, there is still a need to find specific treatments for tumour hypoxia. In this review, we discuss a variety of approaches to alter the low oxygen tumour microenvironment or hypoxia pathways including carbogen breathing, hyperthermia, hypoxia-activated prodrugs, tumour metabolism and hypoxia-inducible factor (HIF) inhibitors. The recent advances in technology and biological understanding reveal the importance of revisiting old therapeutic regimens and repurposing their uses clinically.
Collapse
Affiliation(s)
| | | | - Eui Jung Moon
- Department of Oncology, MRC Oxford Institute for Radiation Oncology, University of Oxford, Headington OX3 7DQ, UK; (N.V.-L.); (M.B.)
| |
Collapse
|
28
|
Staudt M, Jung M. Hypoxia-activated KDAC inhibitor: Taking a breath from untargeted therapy. Cell Chem Biol 2021; 28:1255-1257. [PMID: 34534468 DOI: 10.1016/j.chembiol.2021.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Use of hypoxia-activated prodrugs has emerged as a strategy for selectively targeting tumors in hypoxic conditions harboring reductive environments. In this issue of Cell Chemical Biology, Skwarska et al. (2021) report a hypoxia-activated prodrug targeting histone deacetylases (lysine deacetylases, KDACs) selectively over normoxic cells with activity in an animal model.
Collapse
Affiliation(s)
- Maximilian Staudt
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg, Germany
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg, Germany; Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
29
|
Ellingson BM, Wen PY, Cloughesy TF. Therapeutic Response Assessment of High-Grade Gliomas During Early-Phase Drug Development in the Era of Molecular and Immunotherapies. Cancer J 2021; 27:395-403. [PMID: 34570454 PMCID: PMC8480435 DOI: 10.1097/ppo.0000000000000543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Several new therapeutic strategies have emerged over the past decades to address unmet clinical needs in high-grade gliomas, including targeted molecular agents and various forms of immunotherapy. Each of these strategies requires addressing fundamental questions, depending on the stage of drug development, including ensuring drug penetration into the brain, engagement of the drug with the desired target, biologic effects downstream from the target including metabolic and/or physiologic changes, and identifying evidence of clinical activity that could be expanded upon to increase the likelihood of a meaningful survival benefit. The current review article highlights these strategies and outlines how imaging technology can be used for therapeutic response evaluation in both targeted and immunotherapies in early phases of drug development in high-grade gliomas.
Collapse
Affiliation(s)
- Benjamin M. Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Patrick Y. Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard University, Boston, MA
| | - Timothy F. Cloughesy
- UCLA Neuro Oncology Program, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
30
|
Apilan AG, Mothersill C. Targeted and Non-Targeted Mechanisms for Killing Hypoxic Tumour Cells-Are There New Avenues for Treatment? Int J Mol Sci 2021; 22:8651. [PMID: 34445354 PMCID: PMC8395506 DOI: 10.3390/ijms22168651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 11/25/2022] Open
Abstract
PURPOSE A major issue in radiotherapy is the relative resistance of hypoxic cells to radiation. Historic approaches to this problem include the use of oxygen mimetic compounds to sensitize tumour cells, which were unsuccessful. This review looks at modern approaches aimed at increasing the efficacy of targeting and radiosensitizing hypoxic tumour microenvironments relative to normal tissues and asks the question of whether non-targeted effects in radiobiology may provide a new "target". Novel techniques involve the integration of recent technological advancements such as nanotechnology, cell manipulation, and medical imaging. Particularly, the major areas of research discussed in this review include tumour hypoxia imaging through PET imaging to guide carbogen breathing, gold nanoparticles, macrophage-mediated drug delivery systems used for hypoxia-activate prodrugs, and autophagy inhibitors. Furthermore, this review outlines several features of these methods, including the mechanisms of action to induce radiosensitization, the increased accuracy in targeting hypoxic tumour microenvironments relative to normal tissue, preclinical/clinical trials, and future considerations. CONCLUSIONS This review suggests that the four novel tumour hypoxia therapeutics demonstrate compelling evidence that these techniques can serve as powerful tools to increase targeting efficacy and radiosensitizing hypoxic tumour microenvironments relative to normal tissue. Each technique uses a different way to manipulate the therapeutic ratio, which we have labelled "oxygenate, target, use, and digest". In addition, by focusing on emerging non-targeted and out-of-field effects, new umbrella targets are identified, which instead of sensitizing hypoxic cells, seek to reduce the radiosensitivity of normal tissues.
Collapse
|
31
|
Zhao YQ, Biswas S, Chen Q, Jia M, Zhou Y, Bhuniya S. Direct Readout Hypoxia Tumor Suppression In Vivo through NIR-Theranostic Activation. ACS APPLIED BIO MATERIALS 2021; 4:5686-5694. [PMID: 35006742 DOI: 10.1021/acsabm.1c00467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Urgency in finding a suitable therapy in tumor hypoxia strives to develop hypoxia-targeted activatable theranostic. A strategic theranostic prodrug (Azo-M) has been synthesized. Its azo-linker scission under the hypoxia condition has released an near-infrared (NIR)-reporter to determine the extent of chemotherapeutic (melphalan analogue) activation. Under an artificial hypoxia condition, a large shift from 520 to 590 nm in UV absorption was observed in Azo-M. Alongside, the emission maxima had appeared at 625 nm under the said condition. The Azo-M post-incubated HeLa cells have shown upregulation of various apoptotic factors under oxygen deprivation (3%) condition. Azo-M has shown antiproliferative activity under hypoxia conditions in various cancer cells. An ex-vivo biodistribution study indicated that theranostic Azo-M only activated in tumor tissue and to some extent in the liver. The therapeutic activity study in vivo indicated that Azo-M effectively reduced the tumor size and volume (about 2-fold) without the change of bodyweight of mice. The theranostic Azo-M can be a cornerstone to suppress tumor hypoxia and tracking its extent of suppression.
Collapse
Affiliation(s)
- Yu-Qiang Zhao
- College of Chemical Science and Technology, Yunnan University, Kunming 650091, P. R. China
| | - Shayeri Biswas
- Centre for Interdisciplinary Sciences, JIS Institute of Advanced Studies and Research, JIS University, Kolkata 700091, India
| | - Qiuling Chen
- College of Chemical Science and Technology, Yunnan University, Kunming 650091, P. R. China
| | - Mingxuan Jia
- College of Chemical Science and Technology, Yunnan University, Kunming 650091, P. R. China
| | - Ying Zhou
- College of Chemical Science and Technology, Yunnan University, Kunming 650091, P. R. China
| | - Sankarprasad Bhuniya
- Centre for Interdisciplinary Sciences, JIS Institute of Advanced Studies and Research, JIS University, Kolkata 700091, India
| |
Collapse
|
32
|
Characterization of self-aggregated mitomycin C onto the boron-nitride nanotube as a drug delivery carrier: A molecular dynamics investigation. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
33
|
Omran Z, Guise CP, Chen L, Rauch C, Abdalla AN, Abdullah O, Sindi IA, Fischer PM, Smaill JB, Patterson AV, Liu Y, Wang Q. Design, Synthesis and In-Vitro Biological Evaluation of Antofine and Tylophorine Prodrugs as Hypoxia-Targeted Anticancer Agents. Molecules 2021; 26:3327. [PMID: 34206005 PMCID: PMC8199124 DOI: 10.3390/molecules26113327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 05/20/2021] [Accepted: 05/29/2021] [Indexed: 12/15/2022] Open
Abstract
Phenanthroindolizidines, such as antofine and tylophorine, are a family of natural alkaloids isolated from different species of Asclepiadaceas. They are characterized by interesting biological activities, such as pronounced cytotoxicity against different human cancerous cell lines, including multidrug-resistant examples. Nonetheless, these derivatives are associated with severe neurotoxicity and loss of in vivo activity due to the highly lipophilic nature of the alkaloids. Here, we describe the development of highly polar prodrugs of antofine and tylophorine as hypoxia-targeted prodrugs. The developed quaternary ammonium salts of phenanthroindolizidines showed high chemical and metabolic stability and are predicted to have no penetration through the blood-brain barrier. The designed prodrugs displayed decreased cytotoxicity when tested under normoxic conditions. However, their cytotoxic activity considerably increased when tested under hypoxic conditions.
Collapse
Affiliation(s)
- Ziad Omran
- Department of Pharmaceutical Sciences, Pharmacy Department, Batterjee Medical College, Jeddah 21442, Saudi Arabia
| | - Chris P. Guise
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (C.P.G.); (J.B.S.); (A.V.P.)
| | - Linwei Chen
- State Key Laboratory of Elemento-Organic Chemistry, Research Institute of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China; (L.C.); (Y.L.); (Q.W.)
| | - Cyril Rauch
- School of Veterinary Medicine and Science, University of Nottingham, College Road, Sutton Bonington LE12 5RD, UK;
| | - Ashraf N. Abdalla
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (A.N.A.); (O.A.)
| | - Omeima Abdullah
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (A.N.A.); (O.A.)
| | - Ikhlas A. Sindi
- Department of Biology, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Peter M. Fischer
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Jeff B. Smaill
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (C.P.G.); (J.B.S.); (A.V.P.)
| | - Adam V. Patterson
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (C.P.G.); (J.B.S.); (A.V.P.)
| | - Yuxiu Liu
- State Key Laboratory of Elemento-Organic Chemistry, Research Institute of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China; (L.C.); (Y.L.); (Q.W.)
| | - Qingmin Wang
- State Key Laboratory of Elemento-Organic Chemistry, Research Institute of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China; (L.C.); (Y.L.); (Q.W.)
| |
Collapse
|
34
|
Abi-Jaoudeh N, Dayyani F, Chen PJ, Fernando D, Fidelman N, Javan H, Liang PC, Hwang JI, Imagawa DK. Phase I Trial on Arterial Embolization with Hypoxia Activated Tirapazamine for Unresectable Hepatocellular Carcinoma. J Hepatocell Carcinoma 2021; 8:421-434. [PMID: 34041204 PMCID: PMC8139681 DOI: 10.2147/jhc.s304275] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/20/2021] [Indexed: 01/01/2023] Open
Abstract
Background Tirapazamine (TPZ) is a hypoxia activated drug that may be synergistic with transarterial embolization (TAE). The primary objective was to evaluate the safety of combining TPZ and TAE in patients with unresectable HCC and determine the optimal dose for Phase II. Methods This was a Phase 1 multicenter, open-label, non-randomized trial with a classic 3+3 dose escalation and an expansion cohort in patients with unresectable HCC, Child Pugh A, ECOG 0 or 1. Two initial cohorts consisted of I.V. administration of Tirapazamine followed by superselective TAE while the remaining three cohorts underwent intraarterial administration of Tirapazamine with superselective TAE. Safety and tolerability were assessed using NCI CTCAE 4.0 with clinical, imaging and laboratory examinations including pharmacokinetic (PK) analysis and an electrocardiogram 1 day pre-dose, at 1, 2, 4, 6, 10, and 24 hours post-TPZ infusion and an additional PK at 15- and 30-minutes post-TPZ. Tumor responses were evaluated using mRECIST criteria. Results Twenty-seven patients (mean [range] age of 66.4 [37–79] years) with unresectable HCC were enrolled between July 2015 and January 2018. Two patients were lost to follow-up. Mean tumor size was 6.53 cm ± 2.60 cm with a median of two lesions per patient. Dose limiting toxicity and maximum tolerated dose were not reached. The maximal TPZ dose was 10 mg/m2 I.V. and 20 mg/m2 I.A. One adverse event (AE) was reported in all patients with fatigue, decreased appetite or pain being most common. Grade 3–5 AE were hypertension and transient elevation of AST/ALT in 70.4% of patients. No serious AE were drug related. Sixty percent (95% CI=38.7–78.9) achieved complete response (CR), and 84% (95% CI=63.9–95.5) had complete and partial response per mRECIST for target lesions. Discussion TAE with TPZ was safe and tolerable with encouraging results justifying pursuit of a Phase II trial. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/ZQdL9fUadAE
Collapse
Affiliation(s)
- Nadine Abi-Jaoudeh
- Department of Radiological Sciences, University of California Irvine, Orange, CA, USA
| | - Farshid Dayyani
- Chao Comprehensive Digestive Disease, University of California Irvine, Orange, CA, USA
| | - Pei Jer Chen
- Hepatitis Research Center, National Taiwan University, Taipei City, Taiwan
| | - Dayantha Fernando
- Department of Radiological Sciences, University of California Irvine, Orange, CA, USA
| | - Nicholas Fidelman
- Department of Radiology, University of California San Francisco, San Francisco, CA, USA
| | - Hanna Javan
- Department of Radiological Sciences, University of California Irvine, Orange, CA, USA
| | - Po-Chin Liang
- Department of Medical Imaging, National Taiwan University, Taipei City, Taiwan
| | - Jen-I Hwang
- Department of Radiology, Taichung Veteran General Hospital, Taichung, Taiwan
| | - David K Imagawa
- Surgery Services, University of California Irvine, Orange, CA, USA
| |
Collapse
|
35
|
Fu Z, Mowday AM, Smaill JB, Hermans IF, Patterson AV. Tumour Hypoxia-Mediated Immunosuppression: Mechanisms and Therapeutic Approaches to Improve Cancer Immunotherapy. Cells 2021; 10:1006. [PMID: 33923305 PMCID: PMC8146304 DOI: 10.3390/cells10051006] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 01/05/2023] Open
Abstract
The magnitude of the host immune response can be regulated by either stimulatory or inhibitory immune checkpoint molecules. Receptor-ligand binding between inhibitory molecules is often exploited by tumours to suppress anti-tumour immune responses. Immune checkpoint inhibitors that block these inhibitory interactions can relieve T-cells from negative regulation, and have yielded remarkable activity in the clinic. Despite this success, clinical data reveal that durable responses are limited to a minority of patients and malignancies, indicating the presence of underlying resistance mechanisms. Accumulating evidence suggests that tumour hypoxia, a pervasive feature of many solid cancers, is a critical phenomenon involved in suppressing the anti-tumour immune response generated by checkpoint inhibitors. In this review, we discuss the mechanisms associated with hypoxia-mediate immunosuppression and focus on modulating tumour hypoxia as an approach to improve immunotherapy responsiveness.
Collapse
Affiliation(s)
- Zhe Fu
- Malaghan Institute of Medical Research, Wellington 6042, New Zealand; (Z.F.); (I.F.H.)
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, University of Auckland, Auckland 1142, New Zealand; (A.M.M.); (J.B.S.)
| | - Alexandra M. Mowday
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, University of Auckland, Auckland 1142, New Zealand; (A.M.M.); (J.B.S.)
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Jeff B. Smaill
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, University of Auckland, Auckland 1142, New Zealand; (A.M.M.); (J.B.S.)
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Ian F. Hermans
- Malaghan Institute of Medical Research, Wellington 6042, New Zealand; (Z.F.); (I.F.H.)
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, University of Auckland, Auckland 1142, New Zealand; (A.M.M.); (J.B.S.)
| | - Adam V. Patterson
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, University of Auckland, Auckland 1142, New Zealand; (A.M.M.); (J.B.S.)
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
36
|
Lodhi T, Song YP, West C, Hoskin P, Choudhury A. Hypoxia and its Modification in Bladder Cancer: Current and Future Perspectives. Clin Oncol (R Coll Radiol) 2021; 33:376-390. [PMID: 33762140 DOI: 10.1016/j.clon.2021.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023]
Abstract
Radiotherapy plays an essential role in the curative treatment of muscle-invasive bladder cancer (MIBC). Hypoxia affects the response to MIBC radiotherapy, limiting radiocurability. Likewise, hypoxia influences MIBC genetic instability and malignant progression being associated with metastatic disease and a worse prognosis. Hypoxia identification in MIBC enables treatment stratification and the promise of improved survival. The most promising methods are histopathological markers such as necrosis; biomarkers of protein expression such as HIF-1α, GLUT-1 and CAIX; microRNAs; and novel mRNA signatures. Although hypoxia modification can take different forms, the gold standard remains carbogen and nicotinamide, which improve local control rates in bladder preservation and absolute overall survival with no significant increase in late toxicity. This is an exciting time for evolving therapies such as bioreductive agents, novel oxygen delivery techniques, immunotherapy and poly (ADP-ribose) polymerase 1 (PARP) inhibitors, all in development and representing upcoming trends in MIBC hypoxia modification. Whatever the future holds for hypoxia-modified radiotherapy, there is no doubt of its importance in MIBC. mRNA signatures provide an ideal platform for the selection of those with hypoxic tumours but are yet to qualified and integrated into the clinic. Future interventional trials will require biomarker stratification to ensure optimal treatment response to improve outcomes for patients with MIBC.
Collapse
Affiliation(s)
- T Lodhi
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Y P Song
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - C West
- Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, The Christie NHS Foundation Trust, Manchester, UK
| | - P Hoskin
- Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, The Christie NHS Foundation Trust, Manchester, UK; Cancer Centre, Mount Vernon Hospital, Northwood, UK
| | - A Choudhury
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK; Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, The Christie NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
37
|
Thiruthaneeswaran N, Bibby BAS, Yang L, Hoskin PJ, Bristow RG, Choudhury A, West C. Lost in application: Measuring hypoxia for radiotherapy optimisation. Eur J Cancer 2021; 148:260-276. [PMID: 33756422 DOI: 10.1016/j.ejca.2021.01.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/15/2022]
Abstract
The history of radiotherapy is intertwined with research on hypoxia. There is level 1a evidence that giving hypoxia-targeting treatments with radiotherapy improves locoregional control and survival without compromising late side-effects. Despite coming in and out of vogue over decades, there is now an established role for hypoxia in driving molecular alterations promoting tumour progression and metastases. While tumour genomic complexity and immune profiling offer promise, there is a stronger evidence base for personalising radiotherapy based on hypoxia status. Despite this, there is only one phase III trial targeting hypoxia modification with full transcriptomic data available. There are no biomarkers in routine use for patients undergoing radiotherapy to aid management decisions, and a roadmap is needed to ensure consistency and provide a benchmark for progression to application. Gene expression signatures address past limitations of hypoxia biomarkers and could progress biologically optimised radiotherapy. Here, we review recent developments in generating hypoxia gene expression signatures and highlight progress addressing the challenges that must be overcome to pave the way for their clinical application.
Collapse
Affiliation(s)
- Niluja Thiruthaneeswaran
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.
| | - Becky A S Bibby
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Lingjang Yang
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Peter J Hoskin
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Mount Vernon Cancer Centre, Northwood, UK
| | - Robert G Bristow
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; CRUK Manchester Institute and Manchester Cancer Research Centre, Manchester, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Catharine West
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| |
Collapse
|
38
|
Abdullah NA, Md Hashim NF, Ammar A, Muhamad Zakuan N. An Insight into the Anti-Angiogenic and Anti-Metastatic Effects of Oridonin: Current Knowledge and Future Potential. Molecules 2021; 26:775. [PMID: 33546106 PMCID: PMC7913218 DOI: 10.3390/molecules26040775] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide, with a mortality rate of more than 9 million deaths reported in 2018. Conventional anti-cancer therapy can greatly improve survival however treatment resistance is still a major problem especially in metastatic disease. Targeted anti-cancer therapy is increasingly used with conventional therapy to improve patients' outcomes in advanced and metastatic tumors. However, due to the complexity of cancer biology and metastasis, it is urgent to develop new agents and evaluate the anti-cancer efficacy of available treatments. Many phytochemicals from medicinal plants have been reported to possess anti-cancer properties. One such compound is known as oridonin, a bioactive component of Rabdosia rubescens. Several studies have demonstrated that oridonin inhibits angiogenesis in various types of cancer, including breast, pancreatic, lung, colon and skin cancer. Oridonin's anti-cancer effects are mediated through the modulation of several signaling pathways which include upregulation of oncogenes and pro-angiogenic growth factors. Furthermore, oridonin also inhibits cell migration, invasion and metastasis via suppressing epithelial-to-mesenchymal transition and blocking downstream signaling targets in the cancer metastasis process. This review summarizes the recent applications of oridonin as an anti-angiogenic and anti-metastatic drug both in vitro and in vivo, and its potential mechanisms of action.
Collapse
Affiliation(s)
- Nurul Akmaryanti Abdullah
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Nur Fariesha Md Hashim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Aula Ammar
- Wolfson Wohl Translational Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow City G61 1BD, UK;
| | - Noraina Muhamad Zakuan
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| |
Collapse
|
39
|
Bernauer C, Man YKS, Chisholm JC, Lepicard EY, Robinson SP, Shipley JM. Hypoxia and its therapeutic possibilities in paediatric cancers. Br J Cancer 2021; 124:539-551. [PMID: 33106581 PMCID: PMC7851391 DOI: 10.1038/s41416-020-01107-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 07/20/2020] [Accepted: 09/11/2020] [Indexed: 12/19/2022] Open
Abstract
In tumours, hypoxia-a condition in which the demand for oxygen is higher than its availability-is well known to be associated with reduced sensitivity to radiotherapy and chemotherapy, and with immunosuppression. The consequences of hypoxia on tumour biology and patient outcomes have therefore led to the investigation of strategies that can alleviate hypoxia in cancer cells, with the aim of sensitising cells to treatments. An alternative therapeutic approach involves the design of prodrugs that are activated by hypoxic cells. Increasing evidence indicates that hypoxia is not just clinically significant in adult cancers but also in paediatric cancers. We evaluate relevant methods to assess the levels and extent of hypoxia in childhood cancers, including novel imaging strategies such as oxygen-enhanced magnetic resonance imaging (MRI). Preclinical and clinical evidence largely supports the use of hypoxia-targeting drugs in children, and we describe the critical need to identify robust predictive biomarkers for the use of such drugs in future paediatric clinical trials. Ultimately, a more personalised approach to treatment that includes targeting hypoxic tumour cells might improve outcomes in subgroups of paediatric cancer patients.
Collapse
Affiliation(s)
- Carolina Bernauer
- Sarcoma Molecular Pathology Team, The Institute of Cancer Research, London, UK
| | - Y K Stella Man
- Sarcoma Molecular Pathology Team, The Institute of Cancer Research, London, UK
| | - Julia C Chisholm
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Surrey, UK
- Sarcoma Clinical Trials in Children and Young People Team, The Institute of Cancer Research, London, UK
| | - Elise Y Lepicard
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Simon P Robinson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Janet M Shipley
- Sarcoma Molecular Pathology Team, The Institute of Cancer Research, London, UK.
| |
Collapse
|
40
|
Boyd NH, Tran AN, Bernstock JD, Etminan T, Jones AB, Gillespie GY, Friedman GK, Hjelmeland AB. Glioma stem cells and their roles within the hypoxic tumor microenvironment. Theranostics 2021; 11:665-683. [PMID: 33391498 PMCID: PMC7738846 DOI: 10.7150/thno.41692] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 08/04/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor microenvironments are the result of cellular alterations in cancer that support unrestricted growth and proliferation and result in further modifications in cell behavior, which are critical for tumor progression. Angiogenesis and therapeutic resistance are known to be modulated by hypoxia and other tumor microenvironments, such as acidic stress, both of which are core features of the glioblastoma microenvironment. Hypoxia has also been shown to promote a stem-like state in both non-neoplastic and tumor cells. In glial tumors, glioma stem cells (GSCs) are central in tumor growth, angiogenesis, and therapeutic resistance, and further investigation of the interplay between tumor microenvironments and GSCs is critical to the search for better treatment options for glioblastoma. Accordingly, we summarize the impact of hypoxia and acidic stress on GSC signaling and biologic phenotypes, and potential methods to inhibit these pathways.
Collapse
|
41
|
Lin YH, Satani N, Hammoudi N, Yan VC, Barekatain Y, Khadka S, Ackroyd JJ, Georgiou DK, Pham CD, Arthur K, Maxwell D, Peng Z, Leonard PG, Czako B, Pisaneschi F, Mandal P, Sun Y, Zielinski R, Pando SC, Wang X, Tran T, Xu Q, Wu Q, Jiang Y, Kang Z, Asara JM, Priebe W, Bornmann W, Marszalek JR, DePinho RA, Muller FL. An enolase inhibitor for the targeted treatment of ENO1-deleted cancers. Nat Metab 2020; 2:1413-1426. [PMID: 33230295 PMCID: PMC7744354 DOI: 10.1038/s42255-020-00313-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 10/15/2020] [Indexed: 12/15/2022]
Abstract
Inhibiting glycolysis remains an aspirational approach for the treatment of cancer. We have previously identified a subset of cancers harbouring homozygous deletion of the glycolytic enzyme enolase (ENO1) that have exceptional sensitivity to inhibition of its redundant paralogue, ENO2, through a therapeutic strategy known as collateral lethality. Here, we show that a small-molecule enolase inhibitor, POMHEX, can selectively kill ENO1-deleted glioma cells at low-nanomolar concentrations and eradicate intracranial orthotopic ENO1-deleted tumours in mice at doses well-tolerated in non-human primates. Our data provide an in vivo proof of principle of the power of collateral lethality in precision oncology and demonstrate the utility of POMHEX for glycolysis inhibition with potential use across a range of therapeutic settings.
Collapse
Affiliation(s)
- Yu-Hsi Lin
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nikunj Satani
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute of Stroke and Cerebrovascular Disease, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Naima Hammoudi
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Victoria C Yan
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yasaman Barekatain
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sunada Khadka
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey J Ackroyd
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dimitra K Georgiou
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cong-Dat Pham
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenisha Arthur
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Maxwell
- Institutional Analytics & Informatics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Paul G Leonard
- Core for Biomolecular Structure and Function, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute for Applied Cancer Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Barbara Czako
- Institute for Applied Cancer Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Federica Pisaneschi
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pijus Mandal
- Institute for Applied Cancer Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuting Sun
- Institute for Applied Cancer Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rafal Zielinski
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Susana Castro Pando
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaobo Wang
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Theresa Tran
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Quanyu Xu
- Pharmaceutical Science Facility, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Wu
- Pharmaceutical Science Facility, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yongying Jiang
- Pharmaceutical Science Facility, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhijun Kang
- Institute for Applied Cancer Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John M Asara
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Waldemar Priebe
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William Bornmann
- Director of Drug Discovery and Development, Advanced Organic Synthesis LLC, Houston, Texas, USA
| | - Joseph R Marszalek
- Center for Co-Clinical Trials, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ronald A DePinho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Florian L Muller
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
42
|
Busk M, Overgaard J, Horsman MR. Imaging of Tumor Hypoxia for Radiotherapy: Current Status and Future Directions. Semin Nucl Med 2020; 50:562-583. [PMID: 33059825 DOI: 10.1053/j.semnuclmed.2020.05.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Tumor regions that are transiently or chronically undersupplied with oxygen (hypoxia) and nutrients, and enriched with acidic waste products, are common due to an abnormal and inefficient tumor vasculature, and a deviant highly glycolytic energy metabolism. There is compelling evidence that tumor hypoxia is strongly linked to poor prognosis since oxygen-deprived cells are highly resistant to therapy including radio- and chemotherapy, and survival of such cells is a primary cause of disease relapse. Despite a general improvement in cancer survival rates, hypoxia remains a formidable challenge. Recent progress in radiation delivery systems with improved spatial accuracy that allows dose escalation to hypoxic tumors or even tumor subvolumes, and the development of hypoxia-selective drugs, including bioreductive prodrugs, holds great promise for overcoming this obstacle. However, apart from one notable exception, translation of promising preclinical therapies to the clinic have largely been disappointing. A major obstacle in clinical trials on hypoxia-targeting strategies has been the lack of reliable information on tumor hypoxia, which is crucial for patient stratification into groups of those that are likely to benefit from intervention and those who are not. Further, in many newer trials on hypoxia-selective drugs the choice of cancer disease and combination therapy has not always been ideal, especially not for clinical proof of principle trials. Clearly, there is a pending need for clinical applicable methodologies that may allow us to quantify, map and monitor hypoxia. Molecular imaging may provide the information required for narrowing the gap between potential and actual patient benefit of hypoxia-targeting strategies. The grand majority of preclinical and clinical work has focused on the usefulness of PET-based assessment of hypoxia-selective tracers. Since hypoxia PET has profound inherent weaknesses, the use of other methodologies, including more indirect methods that quantifies blood flow or oxygenation-dependent flux changes through ATP-generating pathways (eg, anaerobic glycolysis) is being extensively studied. In this review, we briefly discuss established and emerging hypoxia-targeting strategies, followed by a more thorough evaluation of strengths and weaknesses of clinical applicable imaging methodologies that may guide timely treatment intensification to overcome hypoxia-driven resistance. Historically, most evidence for the linkage between hypoxia and poor outcome is based on work in the field of radiotherapy. Therefore, main emphasis in this review is on targeting and imaging of hypoxia for improved radiotherapy.
Collapse
Affiliation(s)
- Morten Busk
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital (AUH), Aarhus, Denmark; Danish Centre for Particle Therapy, (AUH), Aarhus, Denmark.
| | - Jens Overgaard
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital (AUH), Aarhus, Denmark
| | - Michael R Horsman
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital (AUH), Aarhus, Denmark
| |
Collapse
|
43
|
The role of single- and multi-walled carbon nanotube in breast cancer treatment. Ther Deliv 2020; 11:653-672. [DOI: 10.4155/tde-2020-0019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Numerous studies have been conducted to design new strategies for breast cancer treatment. Past studies have shown a wide range of carbon-nanomaterials properties, such as single- and multi-walled carbon nanotubes (SWCNTs and MWCNTs) in breast cancer diagnosis and treatment. In this regard, the current study aims to review the role of both SWCNTs and MWCNTs in breast cancer treatment and diagnosis. For reaching this goal, we reviewed the literature by using various searching engines such as Scopus, PubMed, Google Scholar, Web of Science and MEDLINE. This comprehensive review showed that CNTs could dramatically improve breast cancer treatment and could be used as a novel modality to increase diagnostic accuracy; however, no clinical studies have been conducted based on CNTs. In addition, the literature review demonstrates a lack of enough studies to evaluate the side effects of using CNTs.
Collapse
|
44
|
Begunov RS, Sokolov AA, Filimonov SI. Synthesis of Quinone Derivatives of Benzannelated Heterocycles with Bridgehead Nitrogen. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2020. [DOI: 10.1134/s1070428020080084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
45
|
In Silico Identification and Biological Evaluation of Antioxidant Food Components Endowed with IX and XII hCA Inhibition. Antioxidants (Basel) 2020; 9:antiox9090775. [PMID: 32825614 PMCID: PMC7555330 DOI: 10.3390/antiox9090775] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/30/2022] Open
Abstract
The tumor-associated isoenzymes hCA IX and hCA XII catalyze the hydration of carbon dioxide to bicarbonate and protons. These isoforms are highly overexpressed in many types of cancer, where they contribute to the acidification of the tumor environment, promoting tumor cell invasion and metastasis. In this work, in order to identify novel dual hCA IX and XII inhibitors, virtual screening techniques and biological assays were combined. A structure-based virtual screening towards hCA IX and XII was performed using a database of approximately 26,000 natural compounds. The best shared hits were submitted to a thermodynamic analysis and three promising best hits were identified and evaluated in terms of their hCA IX and XII inhibitor activity. In vitro biological assays were in line with the theoretical studies and revealed that syringin, lithospermic acid, and (-)-dehydrodiconiferyl alcohol behave as good hCA IX and hCA XII dual inhibitors.
Collapse
|
46
|
Hamis S, Kohandel M, Dubois LJ, Yaromina A, Lambin P, Powathil GG. Combining hypoxia-activated prodrugs and radiotherapy in silico: Impact of treatment scheduling and the intra-tumoural oxygen landscape. PLoS Comput Biol 2020; 16:e1008041. [PMID: 32745136 PMCID: PMC7425994 DOI: 10.1371/journal.pcbi.1008041] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 08/13/2020] [Accepted: 06/11/2020] [Indexed: 12/30/2022] Open
Abstract
Hypoxia-activated prodrugs (HAPs) present a conceptually elegant approach to not only overcome, but better yet, exploit intra-tumoural hypoxia. Despite being successful in vitro and in vivo, HAPs are yet to achieve successful results in clinical settings. It has been hypothesised that this lack of clinical success can, in part, be explained by the insufficiently stringent clinical screening selection of determining which tumours are suitable for HAP treatments. Taking a mathematical modelling approach, we investigate how tumour properties and HAP-radiation scheduling influence treatment outcomes in simulated tumours. The following key results are demonstrated in silico: (i) HAP and ionising radiation (IR) monotherapies may attack tumours in dissimilar, and complementary, ways. (ii) HAP-IR scheduling may impact treatment efficacy. (iii) HAPs may function as IR treatment intensifiers. (iv) The spatio-temporal intra-tumoural oxygen landscape may impact HAP efficacy. Our in silico framework is based on an on-lattice, hybrid, multiscale cellular automaton spanning three spatial dimensions. The mathematical model for tumour spheroid growth is parameterised by multicellular tumour spheroid (MCTS) data.
Collapse
Affiliation(s)
- Sara Hamis
- School of Mathematics and Statistics, University of St Andrews, St Andrews, Scotland
- Department of Mathematics, College of Science, Swansea University, Swansea, Wales, United Kingdom
| | - Mohammad Kohandel
- Department of Applied Mathematics, University of Waterloo, Waterloo, Canada
| | - Ludwig J. Dubois
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Gibin G. Powathil
- Department of Mathematics, College of Science, Swansea University, Swansea, Wales, United Kingdom
| |
Collapse
|
47
|
Huo D, Jiang X, Hu Y. Recent Advances in Nanostrategies Capable of Overcoming Biological Barriers for Tumor Management. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1904337. [PMID: 31663198 DOI: 10.1002/adma.201904337] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/27/2019] [Indexed: 05/22/2023]
Abstract
Engineered nanomaterials have been extensively employed as therapeutics for tumor management. Meanwhile, the complex tumor niche along with multiple barriers at the cellular level collectively hinders the action of nanomedicines. Here, the advanced strategies that hold promise for overcoming the numerous biological barriers facing nanomedicines are summarized. Starting from tumor entry, methods that promote tissue penetration of nanomedicine and address the hypoxia issue are also highlighted. Then, emphasis is given to the significance of overcoming both physical barriers, such as membrane-associated efflux pumps, and biological features, such as resistance to apoptosis. The pros and cons for an individual approach are presented. In addition, the associated technical problems are discussed, along with the importance of balancing the therapeutic merits and the additional cost of sophisticated nanomedicine designs.
Collapse
Affiliation(s)
- Da Huo
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Xiqun Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Yong Hu
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210093, China
| |
Collapse
|
48
|
Anastassova N, Stoyanov S, Mavrova A, Yancheva D. Spectroscopic and in silico study on the conversion of N,N'-disubstituted hydrazone derivatives of 5-nitrobenzimidazole-2-thione into anion and radical anion products: Implications in hepatotoxicity. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 234:118279. [PMID: 32217448 DOI: 10.1016/j.saa.2020.118279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 03/17/2020] [Indexed: 06/10/2023]
Abstract
The conversion of N,N'-disubstituted hydrazone derivatives of 5-nitrobenzimidazole-2-thione into radical anion and dianion products was studied through infrared (IR) spectroscopy and computational methods. The electrochemical reduction of 3,3'-(5-nitro-2-thioxo-1H-benzo[d]imidazole-1,3(2H)-diyl)bis(N'-(2-methoxybenzylidene))propane-hydrazide was performed directly in the IR cell and the spectral changes were monitored over time in order to identify the spectral bands originating from the reduction product. In order to clarify whether the reduction leads to the generation of radical anion or deprotonated radical dianion, a second spectroscopic experiment was carried out where deprotonation was achieved by treatment with sodium methoxide. Both experiments resulted in distinctly different spectral features, giving evidence that the reduction to radical anion is not accompanied by deprotonation. In order to explain the experimentally observed differences in the hepatotoxicity within the series of N,N'-disubstituted derivatives of 5-nitrobenzimidazole-2-thione, several molecular electronic parameters such as frontier molecular orbitals, spin and charge distribution over fragments, and electron affinities of the studied hydrazone derivatives were compared to those of a previously studied ester derivative. Based on the estimated electronic parameters, it was shown that the type of the side chains (ester, hydrazone etc.) attached to the N-atoms in the nitrobenzimidazole derivatives do not change significantly the propensity of the compounds towards nitro reduction, but however the generated radical anions are characterized by different reactivity accounting for the different hepatotoxicity.
Collapse
Affiliation(s)
- N Anastassova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Build. 9, 1113 Sofia, Bulgaria
| | - S Stoyanov
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Build. 9, 1113 Sofia, Bulgaria
| | - A Mavrova
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria
| | - D Yancheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Build. 9, 1113 Sofia, Bulgaria.
| |
Collapse
|
49
|
Meaney C, Rhebergen S, Kohandel M. In silico analysis of hypoxia activated prodrugs in combination with anti angiogenic therapy through nanocell delivery. PLoS Comput Biol 2020; 16:e1007926. [PMID: 32463836 PMCID: PMC7282674 DOI: 10.1371/journal.pcbi.1007926] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 06/09/2020] [Accepted: 05/05/2020] [Indexed: 01/22/2023] Open
Abstract
Tumour hypoxia is a well-studied phenomenon with implications in cancer progression, treatment resistance, and patient survival. While a clear adverse prognosticator, hypoxia is also a theoretically ideal target for guided drug delivery. This idea has lead to the development of hypoxia-activated prodrugs (HAPs): a class of chemotherapeutics which remain inactive in the body until metabolized within hypoxic regions. In theory, these drugs have the potential for increased tumour selectivity and have therefore been the focus of numerous preclinical studies. Unfortunately, HAPs have had mixed results in clinical trials, necessitating further study in order to harness their therapeutic potential. One possible avenue for the improvement of HAPs is through the selective application of anti angiogenic agents (AAs) to improve drug delivery. Such techniques have been used in combination with other conventional chemotherapeutics to great effect in many studies. A further benefit is theoretically achieved through nanocell administration of the combination, though this idea has not been the subject of any experimental or mathematical studies to date. In the following, a mathematical model is outlined and used to compare the predicted efficacies of separate vs. nanocell administration for AAs and HAPs in tumours. The model is experimentally motivated, both in mathematical form and parameter values. Preliminary results of the model are highlighted throughout which qualitatively agree with existing experimental evidence. The novel prediction of our model is an improvement in the efficacy of AA/HAP combination therapies when administered through nanocells as opposed to separately. While this study specifically models treatment on glioblastoma, similar analyses could be performed for other vascularized tumours, making the results potentially applicable to a range of tumour types. Tumour hypoxia is a well-documented phenomenon with adverse effects for the progression of the cancer. Accordingly, various therapeutic strategies have emerged in recent years to combat its effects. Herein, we present an experimentally-motivated mathematical model used to assess the feasibility of the therapeutic combination of anti angiogenic agents with hypoxia-activated prodrugs. Analysis of the combination therapy shows that delivery through drug nanocells provides the optimal anticancer effect: a novel result which should inspire further examination.
Collapse
Affiliation(s)
- Cameron Meaney
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
- * E-mail:
| | - Sander Rhebergen
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| | - Mohammad Kohandel
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
50
|
Silva VL, Kaassis A, Dehsorkhi A, Koffi CR, Severic M, Abdelhamid M, Nyimanu D, Morris CJ, Al-Jamal WT. Enhanced selectivity, cellular uptake, and in vitro activity of an intrinsically fluorescent copper-tirapazamine nanocomplex for hypoxia targeted therapy in prostate cancer. Biomater Sci 2020; 8:2420-2433. [PMID: 32236169 DOI: 10.1039/c9bm01905g] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In the present work, a copper-tirapazamine (TPZ) nanocomplex [Cu(TPZ)2] was synthesized for selective hypoxia-targeted therapy. The nanocomplex revealed a crystalline form, and exhibited higher lipophilicity, compared to TPZ. Furthermore, its stability was confirmed in different media, with minimum dissociation in serum (∼20% up to 72 h). In contrast to other hypoxia-targeted agents, our intrinsically fluorescent nanocomplex offered an invaluable tool to monitor its cellular uptake and intracellular distribution under both normoxia and hypoxia. The conferred higher cellular uptake of the nanocomplex, especially under hypoxia, and its biocompatible reductive potential resulted in superior hypoxia selectivity in two prostate cancer (PC) cell lines. More promisingly, the nanocomplex showed higher potency in three-dimensional tumor spheroids, compared to TPZ, due to its slower metabolism, and probably deeper penetration in tumor spheroids. Interestingly, the nuclear localization of the intact nanocomplex, combined with its higher DNA binding affinity, as evidenced by the DNA binding assay, resulted in significant S-phase cell-cycle arrest, followed by apoptosis in the three-dimensional spheroid model. In conclusion, the presented findings suggested that the Cu(TPZ)2 nanocomplex can be a promising hypoxia-targeted therapeutic, which could potentiate the efficacy of the existing chemo- and radiotherapy in PC.
Collapse
Affiliation(s)
- Vera L Silva
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | | | | | | | | | | | | | | | | |
Collapse
|